- Review
- Open access
- Published:
Complex regulation of cardiac fibrosis: insights from immune cells and signaling pathways
Journal of Translational Medicine volume 23, Article number: 242 (2025)
Abstract
Cardiac fibrosis is a physiological process that involves the formation of scar tissue in the heart in response to injury or damage. This process is initially a protective measure characterized by enhanced fibroblasts, which are responsible for producing extracellular matrix proteins that provide structural support to the heart. However, when fibrosis becomes excessive, it can lead to adverse outcomes, including increasing tissue stiffness and impaired cardiac function, which can ultimately result in heart failure with a poor prognosis. While fibroblasts are the primary cells involved in cardiac fibrosis, immune cells have also been found to play a vital role in its progression. Recent research has shown that immune cells exert multifaceted effects besides regulation of inflammatory response. Advanced research techniques such as single-cell sequencing and multiomics have provided insights into the specific subsets of immune cells involved in fibrosis and the complex regulation of the process. Targeted immunotherapy against fibrosis is gaining traction as a potential treatment option, but it is still unclear how immune cells achieve this regulation and whether distinct subsets are involved in different roles. To better understand the role of immune cells in cardiac fibrosis, it is essential to examine the classical signaling pathways that are closely related to fibrosis formation. We have also focused on the unique properties of diverse immune cells in cardiac fibrosis and their specific intercommunications. Therefore, this review will delve into the plasticity and heterogeneity of immune cells and their specific roles in cardiac fibrosis, which propose insights to facilitate the development of anti-fibrosis therapeutic strategies.
Background
Although the mortality from cardiovascular disease (CVD) has fluctuated in recent years, cardiac disease remains the leading cause of death and a serious threat to human health [1]. Along with CVD, cardiac fibrosis results from an abnormal reparative programme, such as the excessive extracellular matrix (ECM) deposition and remodelling, impaired matrix metalloproteinase (MMP) suppressive function, and aberrant elevation of infiltrating immune cells, which are present in diverse cardiac diseases including hypertensive heart disease, ischemic heart disease, dilated cardiomyopathy, hypertrophic cardiomyopathy and heart disease related with diabetes or aging [2]. For example, myocardial infarction, a typical ischemic heart disease, is induced by a blockage of blood supply to the heart, with the eventual replacement of dead cardiomyocytes by a fibrotic scar. There are some crucial events characterizing cardiac fibrosis, of which the proliferation and activation of fibroblasts are the key parts. Activated fibroblasts, which undergo myofibroblast transformation, produce and secrete superfluous ECM proteins by initiating a plethora of pro-fibrotic signaling pathways.
Recent studies have manifested that immune cells are involved in the regulation of the pro-fibrotic inflammatory response. Whereas the effect of fibroblasts has been extensively studied, the concrete implications of immune cells on cardiac fibrosis are still unclear. Since the current therapeutic strategies for cardiac fibrosis are unsatisfactory, it is urgent to deepen our comprehensive understanding of immune cells in cardiac fibrosis. Single cell spectrum research has revolutionized our capability to investigate the immune system and explore potential molecular mechanisms [3, 4]. This review will focus on the intricate functions and crosstalk of immune cells in cardiac fibrosis, as well as the well-known fibrotic signaling pathway.
Overview of cardiac fibrosis
Mainly developing in the chronic diseases or repair phase of acute injury, cardiac fibrosis is characterized by the excessive deposition of ECM widely perceived to be composed of collagens I/III, glycoproteins, and proteoglycan [2], which is mediated and secreted by various cells, especially fibroblasts. During normal wound healing procedures, a sophisticated regulating network maintains the balance between ECM protein synthesis and degradation and favors the formation of moderate fibrosis to protect heart from further damage like lethal cardiac capture rupture [5]. Nevertheless, once the balance is broken, aberrant fibrosis destroys and changes the structure and function of heart, also referred to as cardiac remodeling, which leads to elevated stiffness, falling compliance, and ultimately deteriorated systolic and diastolic function [6].
Simply put, cardiac fibrosis can be divided into two groups with distinct pathophysiologic processes, interstitial fibrosis and replacement fibrosis [7, 8]. In the absence of abundant cardiomyocytes loss, interstitial fibrosis appears to alter the ECM composition and the microenvironment of cardiac parenchymal cells, commonly induced by non-ischemic cardiomyopathy such as hypertensive heart disease [8]. Though replacing the necrotic cardiomyocytes with interstitial tissue and cells to preserve the heart integrity, replacement fibrosis is employed to compensate for the defective cardiomyocytes [9] due to the negligible ability of myocardial regeneration in adults [10]. Cardiac fibrosis also significantly impacts the pathophysiology of both heart failure with reduced ejection fraction (HFrEF) and heart failure with preserved ejection fraction (HFpEF), accompanied by some differences. In HFrEF, fibrosis is primarily caused by replacement fibrosis that occurs after the death of cardiomyocytes, often due to ischemic injury such as myocardial infarction or chronic pressure overload. This leads to focal scarring and impaired contractility, contributing to systolic dysfunction [11, 12]. In contrast, in HFpEF, fibrosis is more commonly associated with interstitial and perivascular fibrosis. This type of fibrosis increases myocardial stiffness, leading to diastolic dysfunction. It is frequently linked to systemic inflammation, metabolic stress factors like obesity and diabetes, and endothelial dysfunction [12, 13]. The classification, albeit rough and simple, delineates a deeper understanding of cardiac fibrosis, suggesting unique signaling pathways and cell types are involved in different kinds of fibrosis.
Signaling pathway in cardiac fibrosis
Several key signaling pathways are commonly observed and involved in the pathogenesis of heart fibrosis, regardless of whether it is induced by acute ischemic cardiac disease or prolonged chronic inflammatory injury. These pathways play a significant role in the progression of fibrosis, in addition to the predominant events such as fibroblast phenotypic transition, recruitment of various immune cells, and changes in ECM composition (see Fig. 1). These signaling pathways mediate the expression of numerous interactive genes in fibrosis that not only directly affect ECM secretion, but also indirectly trigger the transformation of diverse cells such as fibroblasts, macrophages, and endothelial cells. A large and growing number of studies have shown that regulation of these signaling pathways contributes significantly to changes in the severity of cardiac fibrosis and cardiac function.
Overview of canonical TGF-β/SMAD and Wnt/β-catenin signaling pathway. Released from TGF-β complex, active TGF-β binding to TGF-β receptor initiates SMAD-dependent signaling pathway and promotes the transcription of profibrotic genes. SMAD6/7 exerts an inhibitory role in TGF-β signaling pathway. Wnt protein binds to receptor (complex receptor comprised of FZD and LRP5/6), which results in instability of destruction complex and subsequent increase of β-catenin. Increased β-catenin translocates into nuclear to enhance targeted profibrotic genes expression. APC adenomatous polyposis coli protein, CK1 casein kinase1, GSK-3β glycogen synthase kinase-3β, LRP5/6 low-density lipoprotein receptor-related protein 5/6, FZD frizzled, TGF-β transforming growth factor-β
TGF-β signaling pathway
Transforming growth factor-β (TGF-β) is one of the most significant cytokines involved in cardiac fibrosis with abundant studies completed. It is the central link of fibrosis formation and is produced by a diverse number of cell types reacting to adverse events, including endothelial cells, epithelial cells, fibroblasts, and immune cells such as macrophages [14]. There are three isoforms of TGF-β, TGF-β1, TGF-β2 and TGF-β3. The TGF-β precursor synthesized in the Endoplasmic reticulum forms dimerization via a disulfide bond and then binds to the latency-associated peptide in secretory vesicle [7]. Latency-associated peptide with mature TGF-β becomes disulfide-linked to latent TGF-β binding protein and the complex is secreted to the ECM in which latency-associated peptide becomes disulfide-linked alternatively to glycoprotein A repetitions predominant (GARP) or leucine rich repeat-containing protein 33 in special cell types and the complex is remain on the cell surface [15, 16]. When injury events happen, to activate latent TGF-β, the latent TGF-β binding protein or GARP complex interacts with various αv integrins on adjacent cells resulting in changes that the complex is cleaved and the active TGF-β is released [17, 18]. But unlike other types of αv integrins, αv β 8 integrins can activate TGF-β signaling pathway through precise Latent TGF-β/ GARP complex without the need for mature TGF-βrelease and diffusion [19].
Signaling is initiated by activated TGF-β through binding to transmembrane serine/threonine kinases, TGF-β type I receptor (TβRI), and TGF-β type II receptor on the cell surface [20]. The signaling is mainly divided into canonical and non-canonical signaling or called SMAD-dependent and SMAD-independent signaling [21]. In the canonical signaling, activated TβRI phosphorylates the receptor-SMAD such as SMAD2, SMAD3, SMAD1, SMAD5, and SMAD8 [20], and promotes active receptor-SMAD binding to SAMD4, forming a trimeric complex which is able to translocate to the nucleus and increase transcription of fibrogenic genes [22]. However, activated TβRI not only phosphorylates the SMADs, but also activates other signalings to modify cell function such as PI3K/AKT, mitogen-activated protein kinase (MAPK) pathways, and JAK/ STAT (known as non-canonical signaling) [23]. In addition, SMAD6 and SMAD7 are inhibitory cytokines that exert suppressive function on receptor-SMAD signaling and SMAD-independent signaling (interact with receptor tyrosine kinase ErbB2 to inhibit the activation of ErbB) to reduce the expression of ECM genes and the transformation of fibroblasts into myofibroblasts [24]. After myocardial infarction, TGF-β/ SMAD3 upregulates the expression of programmed cell death 5, which promotes histone deacetylase 3 ubiquitination to inhibit histone deacetylase 3 and alleviates cardiac fibrosis suggesting programmed cell death 5 is a TGF-β negative feedback cytokine in signaling pathways [25].
TGF-β signaling promotes cardiac fibrosis through a wide range of mechanisms including inducing transformation of fibroblasts into myofibroblasts, cell proliferation, cell apoptosis, induction of epithelial-to-mesenchymal transition, and production of ECM [7, 8, 26]. The pathogenesis of fibrosis caused by TGF-β is diverse and involves many other key cytokines or signaling pathways. Bone morphogenetic Protein 9 inhibits TGF by increasing the level of phosphorylated SMAD1 and limiting SMAD3 activity, thereby attenuating cardiac fibrosis and improving cardiac function [27]. Researches show that protease-activated receptor 2 deletion, non-specific alkaline phosphatase, and NUAK family kinase 1 (one of the members of the AMP-activated protein kinase family) aggravate cardiac fibrosis via accelerating TGF-β signaling with the mechanism that increases collagen secretion by fibroblasts [28,29,30,31]. In contrast, the activation of cardiac fibroblasts is inhibited by high-temperature requirement A serine peptidase 3 and increased expression of TGF-β–induced factor homeobox 1 induced by natriuretic peptide receptor C deletion via degrading TGF-β [32, 33]. Besides, TGF-β has critical effects on the function and phenotype of immune cells, such as recruiting neutrophils and macrophages to injury tissues, instructing the differentiation of Tregs cells and inhibiting antigen presentation capability of dendritic cells [16, 20, 34].
PDGF family
The platelet-derived growth factor (PDGF) family participates in a wide range of pathophysiological activities including the inflammatory reaction and wound healing after cardiac injury, resulting in adverse remodeling and cardiac fibrosis with aberrant regulation [9, 35]. PDGFs are comprised of four distinct monomers (PDGF-A, PDGF-B, PDGF-C, and PDGF-D) and all of them accomplish homodimerization of which PDGF-A and PDGF-B can additionally heterodimerize, leading to five various isoforms (AA, BB, CC, DD, and AB) [36]. The dimer composed of PDGF-A and PDGF-B is activated intracellularly while for PDGF-C and PDGF-D, the dimer is secreted extracellularly as a latent form and later turns into an active state via the process of some proteases or special activators [36]. Activated PDGFs bind to parallel PDGF-receptor (PDGFR) which contains two kinds of monomer (PDGFR-α and PDGFR-β) (see Fig. 2), prompting PDGFR tyrosine kinase to phosphorylate substrates [37] and initiating downstream signaling pathways that involve in diverse fibrogenic proceedings such as myofibroblast differentiation and fibroblast proliferation [38, 39].
The secretion, activation and ligand-receptor binding of PDGF signaling pathway. PDGF-A, and PDGF-B are activated intracellularly while PDGF-C and PDGF-D are secreted in a latent state and activated by matrix enzymes extracellularly. Distinct affinity exists in binding between PDGFs and receptors. PDGF: platelet-derived growth factor
The severity of cardiac fibrosis depends on the expression of PDGF subtypes and their related receptors. In transgenic mice, overexpression of PDGF-A leads to the most severe cardiac fibrosis and subsequent fatal heart failure with the mechanism where cardiac interstitial cells (mainly cardiac fibroblasts) express PDGFR-α and are stimulated to produce extracellular matrix, as PDGF-A interacts to PDGFR-α with the highest affinity [40]. PDGF-B and PDGF-D are implicated to exacerbate cardiac fibrosis whereas the ameliorated cardiac fibrosis occurs in overexpression of PDGF-C [41]. The urokinase plasminogen activator produced by macrophages can activate adipocyte-derived PDGF-D by promoting its homologous dimerization, exacerbating cardiac fibrosis and harmful cardiac remodeling [42]. It is speculated that this pathway is more pronounced in overweight or obese heart disease patients. In a model of dilated cardiomyopathy caused by Lamin A/C mutations constructed in vitro, transcriptomic analysis revealed that the PDGF signaling pathway was elevated, and inhibiting this pathway could improve adverse cell phenotypes [43]. Moreover, a study suggests that the PDGF signaling pathway participates in and promotes the differentiation of cardiac mesenchymal stem cells into myofibroblasts, aggravating cardiac remodeling and fibrosis in the late stage of myocardial infarction models [44]. Blockade of PDGFR activity in vivo can reduce the severity of myocardial fibrosis and hypertrophy in ischemic heart failure models [44]. In early recovery from injury, PDGFR-α promotes the migration and proliferation of fibroblasts by serum response factor to response intranuclearly. While at the late stage of injury recovery, PDGFR-α is suppressed by TGF-β signaling pathway and the downregulation of PDGFR-α promotes the differentiation of fibroblasts into myofibroblasts, revealing that PDGFR-α may play a seemingly opposite role in different stages of injury recovery [45]. However, PDGF-AB effectively represses the differentiation of fibroblasts into myofibroblasts after pig myocardial infarction and this result also appears in human cardiac tissue administrated with PDGF-AB [46]. Therefore, there may be more mechanisms resembling PDGF-AB in the PDGF signaling pathway that inhibit fibrosis formation.
Wnt signaling pathway
Mounting research has demonstrated the Wnt signaling pathway is prominent in the pathological development of cardiac fibrosis. Based on subsequent receptor types and downstream intracellular effector molecules, Wnt signaling is divided into two categories, namely the β-catenin-dependent signaling (canonical) and the β-independent signaling (non-canonical) [47, 48]. After Wnt protein binds to corresponding receptor (complex receptor comprised of frizzled and lipoprotein receptor-related protein 5/6 in canonical versus comprised of frizzled and receptor tyrosine kinase-like orphan receptor1/2 in non-canonical), the β-catenin-dependent signaling, just as its name implies, promotes expression of profibrotic genes via accumulated β-catenin translocating into nucleus as transcription cofactor [47], whereas the β-independent signaling includes two major avenues, planar cell polarity pathway and Ca2+-dependent pathway, which involve the activation of small guanosine triphosphateases, such as ras-related C3 botulinum toxin substrate 1 and ras homolog gene family member A, phospholipase C, and protein kinase C [49].
Studies have implicated that extensive molecule regulation deteriorates cardiac remodeling and fibrosis by overactivating the Wnt / β-catenin signaling pathway in diverse animal models and experimental data of human cells [50,51,52,53,54]. Besides, in mice with cardiac stress overload resulting from trans-aortic constriction, the loss of β-catenin function in tissue-resident cardiac fibroblasts can significantly improve cardiac function and alleviate cardiac fibrosis, but it does not affect the number of activated fibroblasts [55]. Researchers detect that the circulating Wnt5a protein is markedly positively correlated with poor right ventricular function and fibrosis in human dilated cardiomyopathy patients, which has been confirmed in mouse models with the mechanism of the activation of nuclear factor of activated T cells related to non-canonical pathways [56]. Wnt signaling aggravates cardiac fibrosis and remodeling by stimulating Yes-associated protein [57]. However, Deletion of Wls, a targeted gene of Yes-associated protein regulating the communication between cardiomyocytes and cardiac fibroblasts, reduces the regeneration of neonatal heart by inhibiting non-canonical Wnt signaling, leading to deterioration of cardiac function and fibrosis, accompanied by activation of cardiac fibroblasts [58]. The above studies indicate Wnt non-canonical signaling is involved in adverse cardiac remodeling and the formation of cardiac fibrosis, and plays different roles in different growth or repair stages, promoting myocardial proliferation in the neonatal stage and hastening profibrotic response of fibroblasts in the mature stage. In the pathogenesis of cardiac fibrosis, Wnt signaling is affected by TGF-β signaling, the well-known signaling participating in detrimental fibrosis [59, 60]. Recently, with the function of modulating gene expression at the post-transcription level, miRNAs are obtaining increasing attention in the process of cardiac fibrosis. By de-repressing the Wnt signaling, miRNA-29 exacerbates pathological hypertrophy and fibrosis of cardiac cells in a mouse model experienced cardiac pressure overload due to constriction of the thoracic aorta [61]. Furthermore, miRNA-384-5p and miRNA-145 target regulatory factors of certain genes and can significantly inhibit the activation of cardiac fibroblasts and attenuate cardiac fibrosis [62, 63]. Wnt receptors are widely believed to exist in interstitial cells, such as fibroblasts. Under pressure overload conditions, as one of the key receptors of the Wnt signaling, lipoprotein receptor-related protein 6 derived from cardiomyocytes is overexpressed, significantly suppressing the expression of β-catenin, through its interplay with cathepsin D (a protein) that reduces the quantity of Wnt5a and Wnt11, ultimately enhancing cardiac function and alleviating cardiac fibrosis [64]. This study suggests there may be an autocrine or paracrine pathway in the myocardial cell population that regulates the chronic repair process of impaired heart via regulating the Wnt signaling.
Others
Follistatin-like protein 1 (FSTL1) is a secreted glycoprotein widely expressed in various tissues and has been found to be closely related to the occurrence and development of cardiac fibrosis in recent years [65, 66]. The role of FSTL1 in cardiac fibrosis is dual. On one hand, FSTL1 promotes cardiac fibrosis by activating the TGF-β/SMAD3 signaling pathway [67]. Research has demonstrated that the absence of FSTL1 derived from endothelial cells led to abnormal activation of SMAD3, which in turn caused fibrosis in the atrium, venous walls, and heart valves [68]. On the other hand, FSTL1 also contributes positively to myocardial cell proliferation and repair. Some studies have shown that promoting the expression of FSTL1 protected against cardiac remodeling and reduced fibrosis [69, 70]. Moreover, specific knockout of FSTL1 augmented pro-inflammatory response with elevated macrophages and pro-inflammatory cytokines/chemokines [71].
MMPs are a group of zinc-dependent endopeptidases that play a crucial role in the degradation and remodeling of the ECM. Based on their substrate specificity, MMPs can be classified into several categories: collagenases, gelatinases, matrix lysins, and membrane-type MMPs [72]. The activity of MMPs is regulated by tissue inhibitors of MMPs [73]. In cardiac fibrosis, the activities of MMP-2 and MMP-9 are significantly increased, leading to the degradation of basement membrane components, promoting fibroblast activation, and increasing collagen deposition [74, 75]. Therefore, emerging investigations have focused on MMP inhibitors, which effectively attenuated adverse cardiac remodeling [76, 77]. Addressing ECM degradation and inhibiting cardiac fibrosis is essential for preserving cardiac function and reducing adverse remodeling.
Immune cells involved in cardiac fibrosis
In addition to the well-known function of eliminating harmful substances via the immune system, the immune cells also participate in the progression of cardiac fibrosis by promoting the activation of various signaling pathways (see Fig. 3). Abundant experimental evidence delineates that immune cells harbor the ability to mediate cytokine secretion from diverse cell types involved in chronic inflammation in impaired cardiac areas, to facilitate the transition to myofibroblasts, and to regulate the phenotypic transformation of crucial cells related to cardiac fibrosis. Taken together, immune cells play a significant role in cardiac homeostasis where the disruption of balance between injury and reparative process may lead to adverse fibrosis and an increasing number of studies unveils advanced insights describing in-depth mechanisms.
Involvement of immune cells in cardiac fibrosis. After heart injuries, immune cells are recruited to damaged cardiac tissue by chemokines. Activated and transformed immune cells secrete numerous chemokines, cytokines, growth factors and enzymes to intricately regulate fibroblast activation and fibrosis formation. Th helper T cell, Treg regulatory T cell, M1 pro-inflammatory macrophage, M2 anti-inflammatory macrophage, CCL c–c motif chemokine ligand, NETs neutrophil extracellular traps, TGF-β transforming growth factor-β
Lymphocyte
It is well established that lymphocytes are the key members of the adaptive immune system. However, recent research has highlighted their important impact on fibrosis response following heart injury. Lymphocytes consist of T lymphocytes and B lymphocytes and T lymphocytes are divided into CD4 + populations and CD8 + populations. In the aged heart, T cells secrete interferon-γ (IFN-γ) to accelerate cardiac fibrosis and the decline in cardiac function [78], which can be blunted by IFN-γ neutralizing antibodies [79]. Theresa Dolejsi, Thomas Schuetz, et al. identified that T cell-produced IFN-γ mediates inflammation and activation of immune cells to impair the regenerative process after myocardial infarction through adoptive transfer of adult T cells into neonatal heart [80]. The heart regenerative capacity is regulated by substantial backgrounds and complex mechanisms, which can not be explained clearly by a single T cell-related mechanism. Achieving complete heart regeneration may be difficult, but it is prospective to promote beneficial fibrosis and enhance heart function by regulating T cell differentiation phenotype, immune activity, and inflammatory response [81]. Moreover, as a typical proinflammatory cytokine, tumor necrosis factor-α (TNF-α) alleviates deleterious cardiac inflammation and fibrosis by inducing the death of effector CD4 + T cells in myocarditis models [82]. Tregs are considered as key immunosuppressive mediators and protectors of heart repair and fibrosis for their function of inhibiting immoderate inflammatory reaction, dampening immune cell infiltration, and stimulating transformation into protective phenotype [83,84,85]. In the neonatal heart, genes that promote tissue regeneration and regulate innate immune cells such as Apoe, Cxcl4, and Atf3 are upregulated in Tregs, promoting the cardiac regeneration process [84, 86]. Inhibiting interleukin (IL)−35 (a class of immune regulating cytokines mainly secreted by Tregs cells) will impair the heart reparative function after myocardial infarction, increase mortality caused by heart rupture, and exacerbate heart function by reducing the proliferation of Ly6Clow MHCIIlow CCR2-macrophages, repressing the expression of fibrosis-related genes, such as TGF-β activation genes and collagen synthesis genes [87]. Cardiac fibrosis itself is a process of damage repair that plays a protective role, so excessive inhibition of profibrotic process may lead to detrimental outcomes. A study found that inhibition of C–C chemokine ligand (CCL)17 could alleviate cardiac injury and adverse ventricular remodeling by increasing Tregs metastasis and recruitment [88]. Nevertheless, the increased Foxp3 + Tregs in ischemic cardiomyopathy exert pro-inflammatory function and worsen cardiac remodeling and fibrosis while losing their immunosuppressive effect by a blurred mechanism [89]. The aforementioned study indicates that Tregs exhibit varying functions depending on the specific context in which they are found. Further investigation is needed on the specific pathways through which Tregs promote heart tissue repair and the distinct functions of manifold subtypes. In Ang II-induced cardiac fibrosis, miRNA-214, IL-9 and Kruppel like factor 10 are validated as vital regulators of fibrosis responses [90, 91]. Intriguingly, the perturbation of gut dysbiosis induced by cardiac pressure overload affects T cell activation and leukocyte infiltration, leading to cardiac remodeling and fibrosis [92]. Th cells have been identified to promote the transformation into aggravated cardiomyopathy [93, 94] and participate in cardiac fibrosis, which is associated with fibrosis signaling pathways, such as TGF-β/SMADs [95,96,97].
Producing antibody is the hallmark of B cells which could enhance cardiac injury and subsequent fibrosis via an antibody-dependent pathway [98, 99]. In an Ig-deficient model, cardiac impairment induced by myocardial infarction is markedly blunted accompanied by improved heart function [100]. Besides, several studies accentuated the role of the complement system activated by antibodies in cardiac homeostasis including profibrotic procedure [101, 102]. Depletion of B cells inhibits myocardial cell proliferation and cardiac regeneration in neonatal mice after apical resection, promoting the formation of cardiac fibrosis and cardiac dysfunction whereas the left anterior descending coronary ligated adult mice with the same administration produce almost opposite results, namely reduced inflammatory response, attenuated fibrosis and improved cardiac function, which may be related to the reduction of B cells that overexpress S100 calcium binding protein A6 and S100 calcium binding protein A4 [103]. This study indicates that the immune system has undergone significant changes during its development and investigation of how these changes occur may suggest the reasons why heart loses regeneration ability in adulthood. Researchers improved cardiac function and reduced cardiac fibrosis by adaptive transfer of Breg following myocardial infarction where Ly6Chi monocytes were significantly reduced in the infarcted area [104]. Reduction of C–C motif chemokine receptor (CCR)2 in monocytes regulated by Breg inhibits the activation and recruitment of pro-inflammatory cells. In another study utilizing an MI model, B cells produce CCL7 and induce Ly6Chi monocyte mobilization to recruit to damaged heart site, increasing cardiac injury and fibrosis as demonstrated by the experiments of specific deletion of CCL7 in B cells [105]. The study does not elucidate the specific mechanism through which B cells recruit Ly6Chi monocytes and their precise subtypes, and the role of CCL7 deserves further investigation. Activated B cells also have the capability to secrete various cytokines such as IL-10 and TNF-α which may be involved in cell differentiation, modulation of gene expression, and sustained activation of inflammation, resulting in cardiac fibrosis [106,107,108]. Targeted elimination of CD20 + B cells using Rituximab can significantly ameliorate cardiac fibrosis [109].
Macrophage
When the heart gets attacked or undergoes chronic injury, macrophages are one of the first corresponding immune cells. Based on function natures, macrophages are roughly divided into two types, namely M1 (pro-inflammatory) and M2 (anti-inflammatory) [110]. M2 produces and secretes cytokines and circulating RNAs such as IL-10, VSIG4, and circUbe3a, which subsequently alter the biological function of cardiac fibroblasts and initiate fibrotic response [111,112,113]. This effectively controlled pro-fibrotic process mainly plays a protective role. M1 activates fibroblasts through miRNA-21, mediating pressure overload-induced cardiac fibrosis and cardiac dysfunction [114]. But with the application of more advanced technologies, a cognition is emerging that M1 and M2 classifications are the two extreme states among which the roles of various macrophages are affected by the expression of different receptors and ligands [115, 116]. According to the origin, CCR2 + macrophages derive from bone marrow progenitor cells and achieve replenishment via monocyte recruitment mainly promoting inflammation [117], whereas CCR2- macrophages originate in the embryonic development harboring the capacity of self-renewal with the function in cardiac homeostasis [118]. For example, CCL17 secreted by CCR2 + macrophages inhibits Tregs recruitment by binding to CCR4, exacerbating ventricular remodeling and increasing myocardial fibrosis [88]. The disorder of crosstalk between immune cells contributes to the formation of harmful fibrosis. In cardiac ischemia–reperfusion injury, IL-34 activating the NF-κB pathway induces the expression of CCL2, which in turn promotes the polarization of CCR2 + macrophages, leading to deleterious cardiac fibrosis [119]. However, adoptive transfer of M2-like (CCR2 + CD206 +) macrophages can alleviate cardiac toxicity and fibrosis in a non-infarctive heart failure model induced by doxorubicin [120].
Macrophages can form a phenotype like myofibroblasts, expressing marker genes of fibroblasts, such as α-SMA (α-smooth muscle actin), FSP1 (fiber last specific protein-1), and COL1A1 (Collagen type I alpha 1) [121]. Recent studies manifest that affected by diverse cytokines CCR2- macrophages activate cardiac fibroblasts to express collagen and ECM via complex pathways such as TGF-β and NADPH oxidase 4 dependent channel [87, 122, 123]. Through hypoxia inducible factor-1α dependent pathway Ly6Chi macrophages recruit to hypoxic impaired areas and secrete oncostatin-m, a member of the IL-6 family, to inhibit TGF-β/ SMAD pathway [124]. Specific deletion of bone marrow cell WWP2 (an E3 ubiquitin ligase) affects the function of Ly6chi macrophages through the IRF7-Ccl5 axis, inhibits the transdifferentiation of myofibroblasts and ultimately alleviates cardiac fibrosis [125]. The crosstalks between macrophages and fibroblasts depend on various molecules, initiating intracellular fibrosis programs. Targeting these molecules may effectively prevent fibrosis progression. Research found that the deficiency of EGF-like repeats and discoidin I-like domains protein 3 (an endogenous inhibitor of neutrophil adhesion) promoted the recruitment and release of neutrophil extracellular traps (NETs) following myocardial infarction, which facilitated the transition from monocyte to Mertk MHCIIlow macrophages through Toll-like receptor (TLR)9, reducing collagen production and α-SMA + myofibroblasts [126]. Compared with the conventional concept that collagen is completely secreted by myofibroblasts, a study has found that in mouse and zebrafish heart attack models, macrophages can secrete collagen directly promoting fibrosis [127]. Justin F. Deniset et al. revealed an intriguing discovery that pericardium-resident Gata6 + macrophages migrate to the damaged heart area after myocardial infarction which results in phenotype conversion, attenuates cardiac injury and inhibits fibrosis formation while the ablation of Gata6 + pericardial cavity macrophages (GPCMs) does not apparently influence fibrosis extent following myocardial infarction [128, 129]. It is a possible elucidation that transdifferentiation of other macrophage subtypes into GPCMs counteracts the regulation of ablated cells in cardiac fibrosis. At present, it has been found that cardiac macrophages exhibit multiple phenotypes and play different functions. However, it is not fully understood whether this alteration in phenotype proportion is the cause or result relative to the final fibrosis, and the molecular mechanisms involved also require further investigation.
Neutrophil
Before the infiltration of other inflammatory cells, neutrophils are recognized as the first responders to various cardiac damages [130, 131]. Following injuries, such as ischemic cardiomyopathy and myocarditis, neutrophils rich in impaired zones not only secrete multiple proteases to eliminate cell debris but also contribute to the resolution of inflammation and tissue repair where macrophages and neutrophils are transited into a reparative phenotype [132,133,134,135,136]. At hemostasis state, neutrophils mainly explore a protective effect to assist with cardiac restoration. Once the hemostasis is disrupted, aberrant neutrophils in a pathological context produce excessive proinflammatory mediators which may cause damage to heart and lead to maladaptive remodeling [137, 138]. Calcium sensing receptor activates nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 inflammasomes in neutrophils, leading to cardiac fibrosis and ventricular remodeling after acute myocardial infarction [139]. Moreover, increased neutrophil elastase promotes the recruitment of neutrophils and M1 macrophages to heart, enhancing inflammatory damage and fibrosis of non-infarcted myocardium [140]. Excessive histidine catalyzes pro-inflammatory effects on neutrophils, as confirmed by increased reactive oxygen species and NETs, as well as activation of cardiac fibroblasts in histidine decarboxylase deficient mice [141]. Through the positive feedback loop between neutrophils and IL-1β, neutrophils play a profibrotic role accompanied by activation of specific signals to fibroblasts including TGF-β signaling [142].
NETs are a kind of chromatin structures embellished by histones, cytoplasmic and granular proteins [143] and participate in fibrosis formation via promoting the migration and infiltration of neutrophils into heart [133, 144]. Extensive experiments evidences suggest that inhibition of NETs potently dampens the pathogenesis of cardiac fibrosis [145], in which suppressing PAD4 (a class of nuclear molecules that promote NETs expression) and tACPA (therapeutic anti-citrullinated protein antibody) have been implicated to play a beneficial role in many cardiopathy models [146,147,148]. Intriguingly, NETs expand the polarization of Mertk-MHCIIlo−int macrophages through the TLR9 pathway, which inhibits myofibroblasts from expressing collagen I/III and α-SMA [126]. The above studies imply that NETs play various roles in fibrosis depending on the distinct responding microenvironments and injury phases.
Mast cell
Mast cells are well-known as mediators of allergic reactions. Nonetheless, years of profound investigations indicate mast cells are vital in cardiac tissue metabolism and fibrosis development characterized by a variety of granules including tryptase, chymase, cytokines, and growth factors [149,150,151]. The number of mast cells has a pronounced increase in heart following injury [152,153,154], which triggers degranulation and initiates ensuing inflammation. Enhanced mast cells are found in heart correlated with interstitial fibrosis in hypertensive myocardial damage [155]. A study demonstrated that mast cells accelerated right ventricle remodeling and deteriorated heart function induced by pressure overload [156]. A plethora of experimental evidences suggest that chymase is a crucial regulator of cardiac fibrosis through activating fibroblasts like mediating the activation of TGF-β and Ang II [151, 157], as reduced collagen I/III and ameliorated fibrosis occur in the models with blockade of chymase [158,159,160]. Mast cell tryptase also plays a similar role in fibroblasts including promoting proliferation and transition into myofibroblast [161, 162]. In addition, TNF-α and IL-1β derived from mast cells during degranulation lead to adverse remodeling and fibrosis by facilitating detrimental inflammatory response and MMP-9-dependent signaling [163,164,165]. Mast cells have been established to be involved in atrial fibrosis and atrial fibrillation coinciding with elevated PDGF, a pro-fibrotic growth factor [166]. But it is still unclear where the mast cells come from and what the complex mechanism is.
However, mast cells can also exert converse function in specific contexts, in which numerous anti-fibrosis mediators are secreted such as IL-10, IL-13, and CXCL-10, and inhibit ECM deposition and collagen synthesis [151, 167, 168]. Deletion of mast cells in hyperhomocysteinemia-induced cardiac dysfunction results in enhanced perivascular fibrosis with aggravated heart function suggesting a protective role of mast cells [169]. Another study manifests that augmented mast cells found in atrium correspond to mitigated cardiac fibrosis and improved functions [170]. There is also in vivo experimental evidence identifying that transplantation of mast cells into murine ischemic heart significantly improves cardiac function and decreases fibrosis extent [171, 172].
In summary, cardiac mast cells are a double-edged sword and several problems to be solved are proposed from the above contradictory conclusions, partially attributed to the failure to select appropriate animal models [173]. Moreover, the common models of mast cell deficiency require mutations of c-Kit which lead to some physiological defects in mast cells. Therefore, achieving a better understanding of how mast cells manipulate cardiac fibrosis process brings valuable insights into promising treatment.
Dendritic cell
Because of genomic research, dendritic cells (DCs) are comprised of conventional DCs (cDCs) and plasmacytoid DCs (pDCs) [174]. Explored more, cDCs contain two subsets, cDCs1, and cDCs2, mediating the adaptive immune response of CD8 + T cells and CD4 + T cells respectively [175]. Plasmacytoid DCs have been demonstrated to hold the capacity to secrete substantial type I interferon with the elusive molecular basis [176]. Beyond presenting antigens to specific immune cells to initiate adaptive immune response, DCs are also essentially involved in the pathogenesis of plenty of diseases including heart failure and cardiac fibrosis [177, 178].
CD103 + /CD11b + cDCs amplify inflammatory responses leading to detriment after MI, as demonstrated by improved cardiac function and diminished fibrosis area of deletion of cDCs [179]. In mice with aldosterone/mineralocorticoid receptor-dependent cardiovascular impairment induced by aldosterone and high-salt diet, activated mineralocorticoid receptors facilitate neutrophil gelatinase-associated lipocalin expression in DCs, enhancing cardiac hypertrophy and fibrosis [180]. Moreover, emerging studies have established that activated DCs contribute to the development of autoimmune reaction-dependent heart damage via recruiting and activating T cells including CD4 + T cells and cytotoxic CD8 + T cells in heart [181,182,183,184]. However, DCs could exert a beneficial role in the repair phase following MI through modulating inflammatory activity and phenotype shift of immune cells such as tissue-specific Tregs and macrophages, which promotes reparative fibrosis and protects against cardiac rupture [178, 185]. For instance, after myocardial infarction, deletion of DCs exacerbates left ventricular remodeling associated with increased inflammatory mediators like IL-1β, IL-18, and TNF-α, compared to the control group. Meanwhile, macrophage subsets are altered by DCs deletion, namely enhanced pro-inflammatory M1 and reduced anti-inflammatory M2, suggesting an immunomodulatory role of DCs [186]. But the accurate crosstalk between DCs and macrophages during myocardial infarction is still ambiguous. Furthermore, Qian Wang et al. found that the adoptive transfer of tolerogenic DCs significantly raised cardiac function and attenuated fibrosis correlated with a decline of inflammation, probably due to inhibited TLR-4/NF-κB signaling [187]. Given that DCs hold the central regulation of immune system, more investigation should be attached to the interaction between DCs and other immune cells in cardiac diseases.
Treatment of cardiac fibrosis
Recent advances in immunology have highlighted the pivotal role of immune cells, particularly in the development and treatment of cardiac fibrosis. Study have shown that modulating macrophage polarization, such as by enhancing glycogen metabolism using inhibitors, can shift M2 macrophages to an anti-fibrotic M1 phenotype, thereby reducing fibroblast activation and collagen deposition [188]. Invariant natural killer T cells, activated by α-galactosylceramide-pulsed dendritic cells treatment, prevented decline in the left ventricular ejection fraction, accompanied by reduced interstitial fibrosis [189]. In mice with ischemic cardiomyopathy by left coronary artery ligation, selective Tregs ablation alleviated the deterioration of heart function and fibrosis [89], inferring the restoration of normal Tregs function may be a promising avenue of therapeutic immunomodulation.
In addition, IL-1β signaling between CCR2 + macrophages and fibroblasts is a key driver of fibrosis. Inhibiting IL-1β signaling through genetic deletion or monoclonal antibodies has shown promise in reducing fibrosis and improving cardiac function [190].
Cardiac fibrosis presents a significant therapeutic challenge, as current treatments primarily aim to slow disease progression rather than reversing established fibrosis. Traditional therapies, such as angiotensin converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs) (see Table 1), target the renin–angiotensin–aldosterone system to reduce fibrosis and ventricular remodeling [191, 192]. However, their effectiveness is often limited in advanced stages of fibrosis, and they may lead to side effects, such as hypotension or hyperkalemia. Emerging therapies are beginning to address these limitations. Chimeric antigen receptor (CAR) -T cell therapy and CAR-macrophage therapy target fibroblast activation protein to eliminate activated fibroblasts, though challenges such as off-target effects and limited tissue infiltration remain [193, 194]. Nanozyme-based therapies use ATP-responsive nanoparticles to deliver antioxidants and siRNA for precise anti-fibrotic effects, although concerns about systemic toxicity exist [195]. Additionally, modulation of the YTHDF3/FLCN/cPLA2 pathway presents a novel approach by regulating lysosomal function and fibrosis through m6A methylation, though its clinical application requires further validation [196].
While traditional therapies remain the cornerstone of treatment, these emerging strategies hold promise for more targeted and effective interventions. Combining these approaches may yield synergistic benefits, but additional research is needed to overcome current limitations and translate these advancements into clinical practice.
Conclusions
Fibrosis is a normal and beneficial physiological process following a variety of heart injuries, nevertheless, while the balance between formation and degradation is out of control, fibrosis plays a detrimental role in deteriorating tissue compliance and heart function instead of a protective role. The review accentuates the importance of immune cells and signaling pathways in cardiac fibrosis and recapitulates distinct properties that orchestrate the complicated fibrotic responses. Furthermore, the effect of immunomodulation on cardiac fibrosis deserves special attention. It was demonstrated that heart transplant recipients experienced more severe cardiac fibrosis compared to healthy ones both in human and mouse [201, 202]. Cyclosporin A, an effective immunosuppressive agent, significantly lead to detriment in cardiac structure and increased fibrosis [203]. In immunocompromised mice with streptozotocin-induced diabetic cardiomyopathy, depletion of T cells mitigated cardiac fibrosis [204]. As an urgently needed direction in clinical practice, the importance of immunomodulation on cardiac fibrosis has been highlighted. Future investigations need to concentrate on several aspects to make progress in the fight against cardiac fibrosis. First, an immune cell-mediated inflammatory response could reduce the area of acute cardiac injury, but chronic excessive inflammation leads to fibrosis and reduced cardiac compliance. Therefore, it is meaningful to choose different strategies to target pro-inflammatory signals and to maintain optimal regulation of inflammatory intensity during the acute or chronic phase of injury. Second, despite the progress in fibrosis, which shows that the reversal of organ fibrosis is possible [14], current anti-fibrotic therapies are mostly targeted at early fibrosis with unmet results. Targeted immunotherapy using engineered immune cells is one of the research highlights [193, 205]. However, some limitations cannot be ignored in chimeric antigen receptor-T cells against cardiac fibrosis. For example, off-target killing of normal cells outside the heart, such as fibroblasts, results in severe dysfunction of other physiological activities. Third, given the complex background ranging from the injury phase to aging and pathophysiology, more well-defined subtypes of cardiac fibrosis need to be classified in order to achieve better targeted treatment. Finding or creating relevant biomarkers to measure the classification and severity of cardiac fibrosis is a wise move [32, 206]. Taken together, due to the dramatic impact of multiple immune cells, signaling pathways and their intercommunications, targeted immunotherapy, especially multi-target, offers the exciting prospect of slowing down and even reversing the progression of cardiac fibrosis.
Availability of data and materials
Not applicable.
Abbreviations
- ACE:
-
Angiotensin converting enzyme
- ARBs:
-
Angiotensin receptor blockers
- CAR:
-
Chimeric antigen receptor
- CCL:
-
C–C chemokine ligand
- CCR:
-
C–C motif chemokine receptor
- cDCs:
-
Conventional DCs
- CVD:
-
Cardiovascular disease
- DCs:
-
Dendritic cells
- ECM:
-
Excessive extracellular matrix
- FSTL1:
-
Follistatin-like protein 1
- GARP:
-
Glycoprotein A repetitions predominant
- GPCMs:
-
Gata6 + pericardial cavity macrophages
- HFpEF:
-
Heart failure with preserved ejection fraction
- HFrEF:
-
Heart failure with reduced ejection fraction
- IFN-γ:
-
Interferon-γ
- IL:
-
Interleukin
- MAPK:
-
Mitogen-activated protein kinase
- MMP:
-
Matrix metalloproteinase
- NETs:
-
Neutrophil extracellular traps
- pDCs:
-
Plasmacytoid DCs
- PDGF:
-
Platelet-derived growth factor
- PDGFR:
-
PDGF-receptor
- TGF-β:
-
Transforming growth factor-β
- TLR:
-
Toll-like receptor
- TNF-α:
-
Tumor necrosis factor-α
- TβRI:
-
TGF-β type I receptor
References
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, et al. 2024 Heart Disease and Stroke Statistics: A Report of US and global data from the American Heart Association. Circulation. 2024;149:e347–913.
Kong P, Christia P, Frangogiannis NG. The pathogenesis of cardiac fibrosis. Cell Mol Life Sci. 2014;71:549–74.
Bhattacharya M, Ramachandran P. Immunology of human fibrosis. Nat Immunol. 2023;24:1423–33.
Litvinukova M, Talavera-Lopez C, Maatz H, Reichart D, Worth CL, Lindberg EL, et al. Cells of the adult human heart. Nature. 2020;588:466–72.
Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 2014;15:786–801.
Tallquist MD, Molkentin JD. Redefining the identity of cardiac fibroblasts. Nat Rev Cardiol. 2017;14:484–91.
Zhao M, Wang L, Wang M, Zhou S, Lu Y, Cui H, et al. Targeting fibrosis, mechanisms and cilinical trials. Signal Transduct Target Ther. 2022;7:206.
de Boer RA, De Keulenaer G, Bauersachs J, Brutsaert D, Cleland JG, Diez J, et al. Towards better definition, quantification and treatment of fibrosis in heart failure. A scientific roadmap by the Committee of Translational Research of the Heart Failure Association (HFA) of the European Society of Cardiology. Eur J Heart Fail. 2019;21:272–85.
Frangogiannis NG. Cardiac fibrosis. Cardiovasc Res. 2021;117:1450–88.
Aguiar CM, Gawdat K, Legere S, Marshall J, Hassan A, Kienesberger PC, Pulinilkunnil T, Castonguay M, Brunt KR, Legare JF. Fibrosis independent atrial fibrillation in older patients is driven by substrate leukocyte infiltration: diagnostic and prognostic implications to patients undergoing cardiac surgery. J Transl Med. 2019;17:413.
Simmonds SJ, Cuijpers I, Heymans S, Jones EAV. Cellular and molecular differences between HFpEF and HFrEF: a step ahead in an improved pathological understanding. Cells. 2020;9:242.
Mishra S, Kass DA. Cellular and molecular pathobiology of heart failure with preserved ejection fraction. Nat Rev Cardiol. 2021;18:400–23.
Lanzer JD, Wienecke LM, Ramirez Flores RO, Zylla MM, Kley C, Hartmann N, Sicklinger F, Schultz JH, Frey N, Saez-Rodriguez J, Leuschner F. Single-cell transcriptomics reveal distinctive patterns of fibroblast activation in heart failure with preserved ejection fraction. Basic Res Cardiol. 2024;119:1001–28.
Lurje I, Gaisa NT, Weiskirchen R, Tacke F. Mechanisms of organ fibrosis: emerging concepts and implications for novel treatment strategies. Mol Aspects Med. 2023;92: 101191.
Massague J, Sheppard D. TGF-beta signaling in health and disease. Cell. 2023;186:4007–37.
Moreau JM, Velegraki M, Bolyard C, Rosenblum MD, Li Z. Transforming growth factor-beta1 in regulatory T cell biology. Sci Immunol. 2022;7:eabi4613.
Peng D, Fu M, Wang M, Wei Y, Wei X. Targeting TGF-β signal transduction for fibrosis and cancer therapy. Mol Cancer. 2022;21:104.
Frangogiannis NG. Transforming growth factor-beta in myocardial disease. Nat Rev Cardiol. 2022;19:435–55.
Campbell MG, Cormier A, Ito S, Seed RI, Bondesson AJ, Lou J, Marks JD, Baron JL, Cheng Y, Nishimura SL. Cryo-EM reveals integrin-mediated TGF-beta activation without release from latent TGF-beta. Cell. 2020;180(490–501): e416.
Batlle E, Massague J. Transforming growth factor-beta signaling in immunity and cancer. Immunity. 2019;50:924–40.
Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-beta: the master regulator of fibrosis. Nat Rev Nephrol. 2016;12:325–38.
Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-β: the master regulator of fibrosis. Nat Rev Nephrol. 2016;12:325–38.
Finnson KW, Almadani Y, Philip A. Non-canonical (non-SMAD2/3) TGF-β signaling in fibrosis: mechanisms and targets. Semin Cell Dev Biol. 2020;101:115–22.
Humeres C, Shinde AV, Hanna A, Alex L, Hernandez SC, Li R, Chen B, Conway SJ, Frangogiannis NG. Smad7 effects on TGF-beta and ErbB2 restrain myofibroblast activation and protect from postinfarction heart failure. J Clin Invest. 2022;132:e146926.
Weng L, Ye J, Yang F, Jia S, Leng M, Jia B, et al. TGF-beta1/SMAD3 regulates programmed cell death 5 that suppresses cardiac fibrosis post-myocardial infarction by inhibiting HDAC3. Circ Res. 2023;133:237–51.
Su J, Morgani SM, David CJ, Wang Q, Er EE, Huang YH, et al. TGF-beta orchestrates fibrogenic and developmental EMTs via the RAS effector RREB1. Nature. 2020;577:566–71.
Morine KJ, Qiao X, York S, Natov PS, Paruchuri V, Zhang Y, Aronovitz MJ, Karas RH, Kapur NK. Bone morphogenetic protein 9 reduces cardiac fibrosis and improves cardiac function in heart failure. Circulation. 2018;138:513–26.
Friebel J, Weithauser A, Witkowski M, Rauch BH, Savvatis K, Dorner A, et al. Protease-activated receptor 2 deficiency mediates cardiac fibrosis and diastolic dysfunction. Eur Heart J. 2019;40:3318–32.
Zhang T, He X, Caldwell L, Goru SK. NUAK1 promotes organ fibrosis via YAP and TGF-β, smad signaling. Sci Transl Med. 2022;14:eaaz4028.
Cheng X, Wang L, Wen X, Gao L, Li G, Chang G, Qin S, Zhang D. TNAP is a novel regulator of cardiac fibrosis after myocardial infarction by mediating TGF-β/Smads and ERK1/2 signaling pathways. EBioMedicine. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ebiom.2021.103370.
Gao L, Wang LY, Liu ZQ, Jiang D, Wu SY, Guo YQ, et al. TNAP inhibition attenuates cardiac fibrosis induced by myocardial infarction through deactivating TGF-beta1/Smads and activating P53 signaling pathways. Cell Death Dis. 2020;11:44.
Ko T, Nomura S, Yamada S, Fujita K, Fujita T, Satoh M, et al. Cardiac fibroblasts regulate the development of heart failure via Htra3-TGF-beta-IGFBP7 axis. Nat Commun. 2022;13:3275.
Meng L, Lu Y, Wang X, Cheng C. NPRC deletion attenuates cardiac fibrosis in diabetic mice by inhibiting TGF smad. Sci Adv. 2023;9:eadd4222.
Hanna A, Frangogiannis NG. The role of the TGF-beta superfamily in myocardial infarction. Front Cardiovasc Med. 2019;6:140.
Klinkhammer BM, Floege J, Boor P. PDGF in organ fibrosis. Mol Aspects Med. 2018;62:44–62.
Papadopoulos N, Lennartsson J. The PDGF/PDGFR pathway as a drug target. Mol Aspects Med. 2018;62:75–88.
Kazlauskas A. PDGFs and their receptors. Gene. 2017;614:1–7.
Zou X, Tang XY, Qu ZY, Sun ZW, Ji CF, Li YJ, Guo SD. Targeting the PDGF/PDGFR signaling pathway for cancer therapy: a review. Int J Biol Macromol. 2022;202:539–57.
Juhl P, Bondesen S, Hawkins CL, Karsdal MA, Bay-Jensen AC, Davies MJ, Siebuhr AS. Dermal fibroblasts have different extracellular matrix profiles induced by TGF-β, PDGF and IL-6 in a model for skin fibrosis. Sci Rep. 2020;10:17300.
Gallini R, Lindblom P, Bondjers C, Betsholtz C, Andrae J. PDGF-A and PDGF-B induces cardiac fibrosis in transgenic mice. Exp Cell Res. 2016;349:282–90.
Folestad E, Kunath A, Wagsater D. PDGF-C and PDGF-D signaling in vascular diseases and animal models. Mol Aspects Med. 2018;62:1–11.
Cheng YW, Zhang ZB, Lan BD, Lin JR, Chen XH, Kong LR, Xu L, Ruan CC, Gao PJ. PDGF-D activation by macrophage-derived uPA promotes AngII-induced cardiac remodeling in obese mice. J Exp Med. 2021;218:e20210252.
Lee J, Termglinchan V, Diecke S, Itzhaki I, Lam CK, Garg P, et al. Activation of PDGF pathway links LMNA mutation to dilated cardiomyopathy. Nature. 2019;572:335–40.
Hamid T, Xu Y, Ismahil MA, Rokosh G, Jinno M, Zhou G, Wang Q, Prabhu SD. Cardiac mesenchymal stem cells promote fibrosis and remodeling in heart failure: role of PDGF signaling. JACC Basic Transl Sci. 2022;7:465–83.
Yao L, Rathnakar BH, Kwon HR, Sakashita H, Kim JH, Rackley A, Tomasek JJ, Berry WL, Olson LE. Temporal control of PDGFRα regulates the fibroblast-to-myofibroblast transition in wound healing. Cell Rep. 2022;40:111192.
Hume RD, Deshmukh T, Doan T, Shim WJ, Kanagalingam S, Tallapragada V, et al. PDGF-AB reduces myofibroblast differentiation without increasing proliferation after myocardial infarction. JACC Basic Transl Sci. 2023;8:658–74.
Burgy O, Konigshoff M. The WNT signaling pathways in wound healing and fibrosis. Matrix Biol. 2018;68–69:67–80.
Zhang Q, Wang L, Wang S, Cheng H, Xu L, Pei G, Wang Y, Fu C, Jiang Y, He C, Wei Q. Signaling pathways and targeted therapy for myocardial infarction. Signal Transduct Target Therapy. 2022;7:78.
Akoumianakis I, Polkinghorne M, Antoniades C. Non-canonical WNT signalling in cardiovascular disease: mechanisms and therapeutic implications. Nat Rev Cardiol. 2022;19:783–97.
Jeong MH, Kim HJ, Pyun JH, Choi KS, Lee DI, Solhjoo S, O’Rourke B, Tomaselli GF, Jeong DS, Cho H, Kang JS. Cdon deficiency causes cardiac remodeling through hyperactivation of WNT/beta-catenin signaling. Proc Natl Acad Sci U S A. 2017;114:E1345–54.
Wang M, Qian L, Li J, Ming H, Fang L, Li Y, et al. GHSR deficiency exacerbates cardiac fibrosis: role in macrophage inflammasome activation and myofibroblast differentiation. Cardiovasc Res. 2020;116:2091–102.
Yin C, Ye Z, Wu J, Huang C, Pan L, Ding H, et al. Elevated Wnt2 and Wnt4 activate NF-κB signaling to promote cardiac fibrosis by cooperation of Fzd4/2 and LRP6 following myocardial infarction. eBioMedicine. 2021;74:103745.
Nayakanti SR, Friedrich A, Sarode P, Jafari L, Maroli G, Boehm M, et al. Targeting Wnt-ss-Catenin-FOSL signaling ameliorates right ventricular remodeling. Circ Res. 2023;132:1468–85.
Hu HH, Cao G, Wu XQ, Vaziri ND, Zhao YY. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res Rev. 2020;60: 101063.
Xiang FL, Fang M, Yutzey KE. Loss of beta-catenin in resident cardiac fibroblasts attenuates fibrosis induced by pressure overload in mice. Nat Commun. 2017;8:712.
Abraityte A, Lunde IG, Askevold ET, Michelsen AE, Christensen G, Aukrust P, et al. Wnt5a is associated with right ventricular dysfunction and adverse outcome in dilated cardiomyopathy. Sci Rep. 2017;7:3490.
Mia MM, Cibi DM, Ghani SABA, Singh A, Tee N, Sivakumar V, Bogireddi H, Cook SA, Mao J, Singh MK. Loss of Yap/Taz in cardiac fibroblasts attenuates adverse remodelling and improves cardiac function. Cardiovasc Res. 2022;118:1785–804.
Liu S, Tang L, Zhao X, Nguyen B, Heallen TR, Li M, Wang J, Wang J, Martin JF. Yap promotes noncanonical Wnt signals from cardiomyocytes for heart regeneration. Circ Res. 2021;129:782–97.
Blyszczuk P, Muller-Edenborn B, Valenta T, Osto E, Stellato M, Behnke S, et al. Transforming growth factor-beta-dependent Wnt secretion controls myofibroblast formation and myocardial fibrosis progression in experimental autoimmune myocarditis. Eur Heart J. 2017;38:1413–25.
Czepiel M, Diviani D, Jazwa-Kusior A, Tkacz K, Rolski F, Smolenski RT, Siedlar M, Eriksson U, Kania G, Blyszczuk P. Angiotensin II receptor 1 controls profibrotic Wnt/beta-catenin signalling in experimental autoimmune myocarditis. Cardiovasc Res. 2022;118:573–84.
Sassi Y, Avramopoulos P, Ramanujam D, Gruter L, Werfel S, Giosele S, et al. Cardiac myocyte miR-29 promotes pathological remodeling of the heart by activating Wnt signaling. Nat Commun. 2017;8:1614.
Seo HH, Lee S, Lee CY, Lee J, Shin S, Song BW, Kim IK, Choi JW, Lim S, Kim SW, Hwang KC. Multipoint targeting of TGF-beta/Wnt transactivation circuit with microRNA 384–5p for cardiac fibrosis. Cell Death Differ. 2019;26:1107–23.
Cui S, Liu Z, Tao B, Fan S, Pu Y, Meng X, Li D, Xia H, Xu L. miR-145 attenuates cardiac fibrosis through the AKT/GSK-3β/β-catenin signaling pathway by directly targeting SOX9 in fibroblasts. J Cell Biochem. 2021;122:209–21.
Wang X, Zou Y, Chen Z, Li Y, Pan L, Wang Y, et al. Low-density lipoprotein receptor-related protein 6 regulates cardiomyocyte-derived paracrine signaling to ameliorate cardiac fibrosis. Theranostics. 2021;11:1249–68.
El-Armouche A, Ouchi N, Tanaka K, Doros G, Wittköpper K, Schulze T, Eschenhagen T, Walsh K, Sam F. Follistatin-like 1 in chronic systolic heart failure: a marker of left ventricular remodeling. Circ Heart Fail. 2011;4:621–7.
Horak M, Fairweather D, Kokkonen P, Bednar D, Bienertova-Vasku J. Follistatin-like 1 and its paralogs in heart development and cardiovascular disease. Heart Fail Rev. 2022;27:2251–65.
Wang Y, Yu K, Zhao C, Zhou L, Cheng J, Wang DW, Zhao C. Follistatin attenuates myocardial fibrosis in diabetic cardiomyopathy via the TGF-β-Smad3 pathway. Front Pharmacol. 2021;12: 683335.
Jiang H, Zhang L, Liu X, Sun W, Kato K, Chen C, et al. Angiocrine FSTL1 (follistatin-like protein 1) insufficiency leads to atrial and venous wall fibrosis via SMAD3 activation. Arterioscler Thromb Vasc Biol. 2020;40:958–72.
Xiao Y, Zhang Y, Chen Y, Li J, Zhang Z, Sun Y, et al. Inhibition of MicroRNA-9-5p protects against cardiac remodeling following myocardial infarction in mice. Hum Gene Ther. 2019;30:286–301.
Shen H, Cui G, Li Y, Ye W, Sun Y, Zhang Z, et al. Follistatin-like 1 protects mesenchymal stem cells from hypoxic damage and enhances their therapeutic efficacy in a mouse myocardial infarction model. Stem Cell Res Ther. 2019;10:17.
Miyabe M, Ohashi K, Shibata R, Uemura Y, Ogura Y, Yuasa D, et al. Muscle-derived follistatin-like 1 functions to reduce neointimal formation after vascular injury. Cardiovasc Res. 2014;103:111–20.
de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix metalloproteinases: from molecular mechanisms to physiology, pathophysiology, and pharmacology. Pharmacol Rev. 2022;74:712–68.
Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuña JM, Perez-Romero BA, Guerrero-Rodriguez JF, Martinez-Avila N, Martinez-Fierro ML. The roles of matrix metalloproteinases and their inhibitors in human diseases. Int J Mol Sci. 2020;21:9739.
Rodrigues KE, Pontes MHB, Cantão MBS, Prado AF. The role of matrix metalloproteinase-9 in cardiac remodeling and dysfunction and as a possible blood biomarker in heart failure. Pharmacol Res. 2024;206: 107285.
Ribeiro Vitorino T, Ferraz do Prado A, Bruno de Assis Cau S, Rizzi E. MMP-2 and its implications on cardiac function and structure: interplay with inflammation in hypertension. Biochem Pharmacol. 2023;215: 115684.
Chan BYH, Roczkowsky A, Cho WJ, Poirier M, Sergi C, Keschrumrus V, Churko JM, Granzier H, Schulz R. MMP inhibitors attenuate doxorubicin cardiotoxicity by preventing intracellular and extracellular matrix remodelling. Cardiovasc Res. 2021;117:188–200.
Niu H, Liu Z, Guan Y, Wen J, Dang Y, Guan J. Harnessing synergistic effects of MMP-2 Inhibition and bFGF to simultaneously preserve and vascularize cardiac extracellular matrix after myocardial infarction. Acta Biomater. 2025;191:189–204.
Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol. 2004;4:583–94.
Pan X-X, Wu F, Chen X-H, Chen D-R, Chen H-J, Kong L-R, Ruan C-C, Gao P-J. T-cell senescence accelerates angiotensin II-induced target organ damage. Cardiovasc Res. 2021;117:271–83.
Dolejsi T, Delgobo M, Schuetz T, Tortola L, Heinze KG, Hofmann U, et al. Adult T-cells impair neonatal cardiac regeneration. Eur Heart J. 2022;43:2698–709.
Wang J, Seo MJ, Deci MB, Weil BR, Canty JM, Nguyen J. Effect of CCR2 inhibitor-loaded lipid micelles on inflammatory cell migration and cardiac function after myocardial infarction. Int J Nanomed. 2018;13:6441–51.
Rolski F, Tkacz K, Weglarczyk K, Kwiatkowski G, Pelczar P, Jazwa-Kusior A, et al. TNF-alpha protects from exacerbated myocarditis and cardiac death by suppressing expansion of activated heart-reactive CD4+ T cells. Cardiovasc Res. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/cvr/cvad158.
Fung THW, Yang KY, Lui KO. An emerging role of regulatory T-cells in cardiovascular repair and regeneration. Theranostics. 2020;10:8924–38.
Li J, Yang KY, Tam RCY, Chan VW, Lan HY, Hori S, Zhou B, Lui KO. Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner. Theranostics. 2019;9:4324–41.
Zacchigna S, Martinelli V, Moimas S, Colliva A, Anzini M, Nordio A, et al. Paracrine effect of regulatory T cells promotes cardiomyocyte proliferation during pregnancy and after myocardial infarction. Nat Commun. 2018;9:2432.
Li J, Liang C, Yang KY, Huang X, Han MY, Li X, et al. Specific ablation of CD4+ T-cells promotes heart regeneration in juvenile mice. Theranostics. 2020;10:8018–35.
Jia D, Jiang H, Weng X, Wu J, Bai P, Yang W, Wang Z, Hu K, Sun A, Ge J. Interleukin-35 promotes macrophage survival and improves wound healing after myocardial infarction in mice. Circ Res. 2019;124:1323–36.
Feng G, Bajpai G, Ma P, Koenig A, Bredemeyer A, Lokshina I, Lai L, Forster I, Leuschner F, Kreisel D, Lavine KJ. CCL17 aggravates myocardial injury by suppressing recruitment of regulatory T cells. Circulation. 2022;145:765–82.
Bansal SS, Ismahil MA, Goel M, Zhou G, Rokosh G, Hamid T, Prabhu SD. Dysfunctional and proinflammatory regulatory T-lymphocytes are essential for adverse cardiac remodeling in ischemic cardiomyopathy. Circulation. 2019;139:206–21.
Zhuang R, Chen J, Cheng HS, Assa C, Jamaiyar A, Pandey AK, et al. Perivascular fibrosis is mediated by a KLF10-IL-9 signaling axis in CD4+ T cells. Circ Res. 2022;130:1662–81.
Nosalski R, Siedlinski M, Denby L, McGinnigle E, Nowak M, Cat AND, et al. T-cell-derived miRNA-214 mediates perivascular fibrosis in hypertension. Circ Res. 2020;126:988–1003.
Carrillo-Salinas FJ, Anastasiou M, Ngwenyama N, Kaur K, Tai A, Smolgovsky SA, Jetton D, Aronovitz M, Alcaide P. Gut dysbiosis induced by cardiac pressure overload enhances adverse cardiac remodeling in a T cell-dependent manner. Gut Microbes. 2020;12:1–20.
Walker JA, McKenzie ANJ. T(H)2 cell development and function. Nat Rev Immunol. 2018;18:121–33.
Song J, Chen X, Cheng L, Rao M, Chen K, Zhang N, Meng J, Li M, Liu ZQ, Yang PC. Vitamin D receptor restricts T helper 2-biased inflammation in the heart. Cardiovasc Res. 2018;114:870–9.
Nevers T, Salvador AM, Velazquez F, Ngwenyama N, Carrillo-Salinas FJ, Aronovitz M, Blanton RM, Alcaide P. Th1 effector T cells selectively orchestrate cardiac fibrosis in nonischemic heart failure. J Exp Med. 2017;214:3311–29.
Wang X, Ni L, Wan S, Zhao X, Ding X, Dejean A, Dong C. Febrile temperature critically controls the differentiation and pathogenicity of T helper 17 cells. Immunity. 2020;52:328-341.e325.
Eyerich S, Eyerich K, Pennino D, Carbone T, Nasorri F, Pallotta S, et al. Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. J Clin Invest. 2009;119:3573–85.
Youker KA, Assad-Kottner C, Cordero-Reyes AM, Trevino AR, Flores-Arredondo JH, Barrios R, Fernandez-Sada E, Estep JD, Bhimaraj A, Torre-Amione G. High proportion of patients with end-stage heart failure regardless of aetiology demonstrates anti-cardiac antibody deposition in failing myocardium: humoral activation, a potential contributor of disease progression. Eur Heart J. 2014;35:1061–8.
van den Hoogen P, de Jager SCA, Huibers MMH, Schoneveld AH, Puspitasari YM, Valstar GB, et al. Increased circulating IgG levels, myocardial immune cells and IgG deposits support a role for an immune response in pre- and end-stage heart failure. J Cell Mol Med. 2019;23:7505–16.
Keppner L, Heinrichs M, Rieckmann M, Demengeot J, Frantz S, Hofmann U, Ramos G. Antibodies aggravate the development of ischemic heart failure. Am J Physiol Heart Circ Physiol. 2018;315:H1358-h1367.
Yu M, Wen S, Wang M, Liang W, Li HH, Long Q, Guo HP, Liao YH, Yuan J. TNF-α-secreting B cells contribute to myocardial fibrosis in dilated cardiomyopathy. J Clin Immunol. 2013;33:1002–8.
Zhang C, Li Y, Wang C, Wu Y, Du J. Antagonist of C5aR prevents cardiac remodeling in angiotensin II-induced hypertension. Am J Hypertens. 2014;27:857–64.
Tan Y, Duan X, Wang B, Liu X, Zhan Z. Murine neonatal cardiac B cells promote cardiomyocyte proliferation and heart regeneration. NPJ Regen Med. 2023;8:7.
Jiao J, He S, Wang Y, Lu Y, Gu M, Li D, et al. Regulatory B cells improve ventricular remodeling after myocardial infarction by modulating monocyte migration. Basic Res Cardiol. 2021;116:46.
Zouggari Y, Ait-Oufella H, Bonnin P, Simon T, Sage AP, Guerin C, et al. B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction. Nat Med. 2013;19:1273–80.
Wu L, Dalal R, Cao CD, Postoak JL, Yang G, Zhang Q, Wang Z, Lal H, Van Kaer L. IL-10-producing B cells are enriched in murine pericardial adipose tissues and ameliorate the outcome of acute myocardial infarction. Proc Natl Acad Sci U S A. 2019;116:21673–84.
Zouggari Y, Ait-Oufella H, Bonnin P, Simon T, Sage AP, Guérin C, et al. B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction. Nat Med. 2013;19:1273–80.
Chan CT, Sobey CG, Lieu M, Ferens D, Kett MM, Diep H, et al. Obligatory role for B cells in the development of angiotensin II-dependent hypertension. Hypertension. 2015;66:1023–33.
Ma X-L, Lin Q-Y, Wang L, Xie X, Zhang Y-L, Li H-H. Rituximab prevents and reverses cardiac remodeling by depressing B cell function in mice. Biomed Pharmacotherapy. 2019;114:112348.
Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol. 2013;229:176–85.
Chen B, Huang S, Su Y, Wu YJ, Hanna A, Brickshawana A, Graff J, Frangogiannis NG. Macrophage Smad3 protects the infarcted heart, stimulating phagocytosis and regulating inflammation. Circ Res. 2019;125:55–70.
Wang Y, Li C, Zhao R, Qiu Z, Shen C, Wang Z, Liu W, Zhang W, Ge J, Shi B. CircUbe3a from M2 macrophage-derived small extracellular vesicles mediates myocardial fibrosis after acute myocardial infarction. Theranostics. 2021;11:6315–33.
Wang Y, Zhang Y, Li J, Li C, Zhao R, Shen C, Liu W, Rong J, Wang Z, Ge J, Shi B. Hypoxia induces M2 macrophages to express VSIG4 and mediate cardiac fibrosis after myocardial infarction. Theranostics. 2023;13:2192–209.
Ramanujam D, Schon AP, Beck C, Vaccarello P, Felician G, Dueck A, Esfandyari D, Meister G, Meitinger T, Schulz C, Engelhardt S. MicroRNA-21-dependent macrophage-to-fibroblast signaling determines the cardiac response to pressure overload. Circulation. 2021;143:1513–25.
Li Y, Feng J, Song S, Li H, Yang H, Zhou B, et al. gp130 Controls cardiomyocyte proliferation and heart regeneration. Circulation. 2020;142:967–82.
Fan Q, Tao R, Zhang H, Xie H, Lu L, Wang T, et al. Dectin-1 contributes to myocardial ischemia/reperfusion injury by regulating macrophage polarization and neutrophil infiltration. Circulation. 2019;139:663–78.
Patel B, Bansal SS, Ismahil MA, Hamid T, Rokosh G, Mack M, Prabhu SD. CCR2(+) monocyte-derived infiltrating macrophages are required for adverse cardiac remodeling during pressure overload. JACC Basic Transl Sci. 2018;3:230–44.
Epelman S, Lavine KJ, Beaudin AE, Sojka DK, Carrero JA, Calderon B, et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity. 2014;40:91–104.
Zhuang L, Zong X, Yang Q, Fan Q, Tao R. Interleukin-34-NF-kappaB signaling aggravates myocardial ischemic/reperfusion injury by facilitating macrophage recruitment and polarization. EBioMedicine. 2023;95: 104744.
Liu Y, Wu M, Zhong C, Xu B, Kang L. M2-like macrophages transplantation protects against the doxorubicin-induced heart failure via mitochondrial transfer. Biomater Res. 2022;26:14.
Haider N, Bosca L, Zandbergen HR, Kovacic JC, Narula N, Gonzalez-Ramos S, et al. Transition of macrophages to fibroblast-like cells in healing myocardial infarction. J Am Coll Cardiol. 2019;74:3124–35.
Vendrov AE, Xiao H, Lozhkin A, Hayami T, Hu G, Brody MJ, Sadoshima J, Zhang Y-Y, Runge MS, Madamanchi NR. Cardiomyocyte NOX4 regulates resident macrophage-mediated inflammation and diastolic dysfunction in stress cardiomyopathy. Redox Biol. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.redox.2023.102937.
Revelo XS, Parthiban P, Chen C, Barrow F, Fredrickson G, Wang H, Yucel D, Herman A, van Berlo JH. Cardiac resident macrophages prevent fibrosis and stimulate angiogenesis. Circ Res. 2021;129:1086–101.
Abe H, Takeda N, Isagawa T, Semba H, Nishimura S, Morioka MS, et al. Macrophage hypoxia signaling regulates cardiac fibrosis via Oncostatin M. Nat Commun. 2019;10:2824.
Chen H, Chew G, Devapragash N, Loh JZ, Huang KY, Guo J, et al. The E3 ubiquitin ligase WWP2 regulates pro-fibrogenic monocyte infiltration and activity in heart fibrosis. Nat Commun. 2022;13:7375.
Wei X, Zou S, Xie Z, Wang Z, Huang N, Cen Z, et al. EDIL3 deficiency ameliorates adverse cardiac remodelling by neutrophil extracellular traps (NET)-mediated macrophage polarization. Cardiovasc Res. 2022;118:2179–95.
Simoes FC, Cahill TJ, Kenyon A, Gavriouchkina D, Vieira JM, Sun X, et al. Macrophages directly contribute collagen to scar formation during zebrafish heart regeneration and mouse heart repair. Nat Commun. 2020;11:600.
Deniset JF, Belke D, Lee WY, Jorch SK, Deppermann C, Hassanabad AF, et al. Gata6(+) pericardial cavity macrophages relocate to the injured heart and prevent cardiac fibrosis. Immunity. 2019;51(131–140): e135.
Jin H, Liu K, Huang X, Huo H, Mou J, Qiao Z, He B, Zhou B. Genetic lineage tracing of pericardial cavity macrophages in the injured heart. Circ Res. 2022;130:1682–97.
Okyere AD, Tilley DG. Leukocyte-dependent regulation of cardiac fibrosis. Front Physiol. 2020;11:301.
Zaidi Y, Aguilar EG, Troncoso M, Ilatovskaya DV, DeLeon-Pennell KY. Immune regulation of cardiac fibrosis post myocardial infarction. Cell Signal. 2021;77: 109837.
Horckmans M, Ring L, Duchene J, Santovito D, Schloss MJ, Drechsler M, Weber C, Soehnlein O, Steffens S. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J. 2017;38:187–97.
Weckbach LT, Grabmaier U, Uhl A, Gess S, Boehm F, Zehrer A, et al. Midkine drives cardiac inflammation by promoting neutrophil trafficking and NETosis in myocarditis. J Exp Med. 2019;216:350–68.
Daseke MJ 2nd, Valerio FM, Kalusche WJ, Ma Y, DeLeon-Pennell KY, Lindsey ML. Neutrophil proteome shifts over the myocardial infarction time continuum. Basic Res Cardiol. 2019;114:37.
Ma Y, Yabluchanskiy A, Iyer RP, Cannon PL, Flynn ER, Jung M, Henry J, Cates CA, Deleon-Pennell KY, Lindsey ML. Temporal neutrophil polarization following myocardial infarction. Cardiovasc Res. 2016;110:51–61.
Kain V, Ingle KA, Kabarowski J, Barnes S, Limdi NA, Prabhu SD, Halade GV. Genetic deletion of 12/15 lipoxygenase promotes effective resolution of inflammation following myocardial infarction. J Mol Cell Cardiol. 2018;118:70–80.
Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ Res. 2016;119:91–112.
Liu XH, Pan LL, Deng HY, Xiong QH, Wu D, Huang GY, Gong QH, Zhu YZ. Leonurine (SCM-198) attenuates myocardial fibrotic response via inhibition of NADPH oxidase 4. Free Radic Biol Med. 2013;54:93–104.
Ren Z, Yang K, Zhao M, Liu W, Zhang X, Chi J, Shi Z, Zhang X, Fu Y, Liu Y, Yin X. Calcium-sensing receptor on neutrophil promotes myocardial apoptosis and fibrosis after acute myocardial infarction via NLRP3 inflammasome activation. Can J Cardiol. 2020;36:893–905.
Ogura Y, Tajiri K, Murakoshi N, Xu D, Yonebayashi S, Li S, et al. Neutrophil elastase deficiency ameliorates myocardial injury post myocardial infarction in mice. Int J Mol Sci. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms22020722.
Zhang Z, Ding S, Wang Z, Zhu X, Zhou Z, Zhang W, Yang X, Ge J. Prmt1 upregulated by Hdc deficiency aggravates acute myocardial infarction via NETosis. Acta Pharm Sin B. 2022;12:1840–55.
Chowkwale M, Lindsey ML, Saucerman JJ. Intercellular model predicts mechanisms of inflammation-fibrosis coupling after myocardial infarction. J Physiol. 2023;601:2635–54.
Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–5.
Sorvillo N, Cherpokova D, Martinod K, Wagner DD. Extracellular DNA NET-works with dire consequences for health. Circ Res. 2019;125:470–88.
Zhang XL, Wang TY, Chen Z, Wang HW, Yin Y, Wang L, Wang Y, Xu B, Xu W. HMGB1-promoted neutrophil extracellular traps contribute to cardiac diastolic dysfunction in mice. J Am Heart Assoc. 2022;11: e023800.
Chirivi RGS, van Rosmalen JWG, van der Linden M, Euler M, Schmets G, Bogatkevich G, et al. Therapeutic ACPA inhibits NET formation: a potential therapy for neutrophil-mediated inflammatory diseases. Cell Mol Immunol. 2021;18:1528–44.
Van Bruggen S, Kraisin S, Van Wauwe J, Bomhals K, Stroobants M, Carai P, Frederix L, Van De Bruaene A, Witsch T, Martinod K. Neutrophil peptidylarginine deiminase 4 is essential for detrimental age-related cardiac remodelling and dysfunction in mice. Philos Trans R Soc Lond B Biol Sci. 2023;378:20220475.
Wong SL, Wagner DD. Peptidylarginine deiminase 4: a nuclear button triggering neutrophil extracellular traps in inflammatory diseases and aging. FASEB J. 2018;32:201800691.
Somasundaram P, Ren G, Nagar H, Kraemer D, Mendoza L, Michael LH, Caughey GH, Entman ML, Frangogiannis NG. Mast cell tryptase may modulate endothelial cell phenotype in healing myocardial infarcts. J Pathol. 2005;205:102–11.
Levick SP, Widiapradja A. Mast cells: key contributors to cardiac fibrosis. Int J Mol Sci. 2018;19:231.
Legere SA, Haidl ID, Légaré J-F, Marshall JS. Mast cells in cardiac fibrosis: new insights suggest opportunities for intervention. Front Immunol. 2019;10:580.
Akgul A, Skrabal CA, Thompson LO, Loebe M, Lafuente JA, Noon GP, Youker KA. Role of mast cells and their mediators in failing myocardium under mechanical ventricular support. J Heart Lung Transplant. 2004;23:709–15.
Cimini M, Fazel S, Zhuo S, Xaymardan M, Fujii H, Weisel RD, Li RK. c-kit dysfunction impairs myocardial healing after infarction. Circulation. 2007;116:I77-82.
Ngkelo A, Richart A, Kirk JA, Bonnin P, Vilar J, Lemitre M, et al. Mast cells regulate myofilament calcium sensitization and heart function after myocardial infarction. J Exp Med. 2016;213:1353–74.
Kotov G, Landzhov B, Stamenov N, Stanchev S, Iliev A. Changes in the number of mast cells, expression of fibroblast growth factor-2 and extent of interstitial fibrosis in established and advanced hypertensive heart disease. Annf Anat Anatomischer Anzeiger. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aanat.2020.151564.
Sydykov A, Luitel H, Mamazhakypov A, Wygrecka M, Pradhan K, Pak O, et al. Genetic deficiency and pharmacological stabilization of mast cells ameliorate pressure overload-induced maladaptive right ventricular remodeling in mice. Int J Mol Sci. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21239099.
Jin J, Jiang Y, Chakrabarti S, Su Z. Cardiac mast cells: a two-head regulator in cardiac homeostasis and pathogenesis following injury. Front Immunol. 2022;13: 963444.
Wang Y, Liu CL, Fang W, Zhang X, Yang C, Li J, et al. Deficiency of mouse mast cell protease 4 mitigates cardiac dysfunctions in mice after myocardium infarction. Biochim Biophys Acta Mol Basis Dis. 2019;1865:1170–81.
Matsumoto C, Hayashi T, Kitada K, Yamashita C, Miyamura M, Mori T, et al. Chymase plays an important role in left ventricular remodeling induced by intermittent hypoxia in mice. Hypertension. 2009;54:164–71.
Matsumoto T, Wada A, Tsutamoto T, Ohnishi M, Isono T, Kinoshita M. Chymase inhibition prevents cardiac fibrosis and improves diastolic dysfunction in the progression of heart failure. Circulation. 2003;107:2555–8.
Gailit J, Marchese MJ, Kew RR, Gruber BL. The differentiation and function of myofibroblasts is regulated by mast cell mediators. J Invest Dermatol. 2001;117:1113–9.
Garbuzenko E, Nagler A, Pickholtz D, Gillery P, Reich R, Maquart FX, Levi-Schaffer F. Human mast cells stimulate fibroblast proliferation, collagen synthesis and lattice contraction: a direct role for mast cells in skin fibrosis. Clin Exp Allergy. 2002;32:237–46.
Duerrschmid C, Crawford JR, Reineke E, Taffet GE, Trial J, Entman ML, Haudek SB. TNF receptor 1 signaling is critically involved in mediating angiotensin-II-induced cardiac fibrosis. J Mol Cell Cardiol. 2013;57:59–67.
Kleinbongard P, Schulz R, Heusch G. TNFα in myocardial ischemia/reperfusion, remodeling and heart failure. Heart Fail Rev. 2011;16:49–69.
Fairweather D, Frisancho-Kiss S, Yusung SA, Barrett MA, Davis SE, Gatewood SJ, Njoku DB, Rose NR. Interferon-gamma protects against chronic viral myocarditis by reducing mast cell degranulation, fibrosis, and the profibrotic cytokines transforming growth factor-beta 1, interleukin-1 beta, and interleukin-4 in the heart. Am J Pathol. 2004;165:1883–94.
Liao CH, Akazawa H, Tamagawa M, Ito K, Yasuda N, Kudo Y, et al. Cardiac mast cells cause atrial fibrillation through PDGF-A-mediated fibrosis in pressure-overloaded mouse hearts. J Clin Invest. 2010;120:242–53.
Kologrivova I, Shtatolkina M, Suslova T, Ryabov V. Cells of the immune system in cardiac remodeling: main players in resolution of inflammation and repair after myocardial infarction. Front Immunol. 2021;12: 664457.
Verma SK, Garikipati VNS, Krishnamurthy P, Schumacher SM, Grisanti LA, Cimini M, et al. Interleukin-10 inhibits bone marrow fibroblast progenitor cell-mediated cardiac fibrosis in pressure-overloaded myocardium. Circulation. 2017;136:940–53.
Joseph J, Kennedy RH, Devi S, Wang J, Joseph L, Hauer-Jensen M. Protective role of mast cells in homocysteine-induced cardiac remodeling. Am J Physiol Heart Circ Physiol. 2005;288:H2541-2545.
Legere SA, Haidl ID, Castonguay MC, Brunt KR, Légaré J-F, Marshall JS. Increased mast cell density is associated with decreased fibrosis in human atrial tissue. J Mol Cell Cardiol. 2020;149:15–26.
Shao Z, Nazari M, Guo L, Li SH, Sun J, Liu SM, Yuan HP, Weisel RD, Li RK. The cardiac repair benefits of inflammation do not persist: evidence from mast cell implantation. J Cell Mol Med. 2015;19:2751–62.
Nazari M, Ni NC, Lüdke A, Li SH, Guo J, Weisel RD, Li RK. Mast cells promote proliferation and migration and inhibit differentiation of mesenchymal stem cells through PDGF. J Mol Cell Cardiol. 2016;94:32–42.
Bradding P, Pejler G. The controversial role of mast cells in fibrosis. Immunol Rev. 2018;282:198–231.
Anderson DA 3rd, Dutertre CA, Ginhoux F, Murphy KM. Genetic models of human and mouse dendritic cell development and function. Nat Rev Immunol. 2021;21:101–15.
Vu Manh TP, Bertho N, Hosmalin A, Schwartz-Cornil I, Dalod M. Investigating evolutionary conservation of dendritic cell subset identity and functions. Front Immunol. 2015;6:260.
Reizis B. Plasmacytoid dendritic cells: development, regulation, and function. Immunity. 2019;50:37–50.
Durai V, Murphy KM. Functions of murine dendritic cells. Immunity. 2016;45:719–36.
Nagai T, Honda S, Sugano Y, Matsuyama TA, Ohta-Ogo K, Asaumi Y, et al. Decreased myocardial dendritic cells is associated with impaired reparative fibrosis and development of cardiac rupture after myocardial infarction in humans. J Am Heart Assoc. 2014;3: e000839.
Lee JS, Jeong SJ, Kim S, Chalifour L, Yun TJ, Miah MA, et al. Conventional dendritic cells impair recovery after myocardial infarction. J Immunol. 2018;201:1784–98.
Araos P, Prado C, Lozano M, Figueroa S, Espinoza A, Berger T, Mak TW, Jaisser F, Pacheco R, Michea L, Amador CA. Dendritic cells are crucial for cardiovascular remodeling and modulate neutrophil gelatinase-associated lipocalin expression upon mineralocorticoid receptor activation. J Hypertens. 2019;37:1482–92.
Forte E, Perkins B, Sintou A, Kalkat HS, Papanikolaou A, Jenkins C, et al. Cross-priming dendritic cells exacerbate immunopathology after ischemic tissue damage in the heart. Circulation. 2021;143:821–36.
Ganguly D, Haak S, Sisirak V, Reizis B. The role of dendritic cells in autoimmunity. Nat Rev Immunol. 2013;13:566–77.
Eriksson U, Ricci R, Hunziker L, Kurrer MO, Oudit GY, Watts TH, Sonderegger I, Bachmaier K, Kopf M, Penninger JM. Dendritic cell-induced autoimmune heart failure requires cooperation between adaptive and innate immunity. Nat Med. 2003;9:1484–90.
Eriksson U, Kurrer MO, Sonderegger I, Iezzi G, Tafuri A, Hunziker L, Suzuki S, Bachmaier K, Bingisser RM, Penninger JM, Kopf M. Activation of dendritic cells through the interleukin 1 receptor 1 is critical for the induction of autoimmune myocarditis. J Exp Med. 2003;197:323–31.
Choo EH, Lee J-H, Park E-H, Park HE, Jung N-C, Kim T-H, et al. Infarcted myocardium-primed dendritic cells improve remodeling and cardiac function after myocardial infarction by modulating the regulatory T cell and macrophage polarization. Circulation. 2017;135:1444–57.
Anzai A, Anzai T, Nagai S, Maekawa Y, Naito K, Kaneko H, et al. Regulatory role of dendritic cells in postinfarction healing and left ventricular remodeling. Circulation. 2012;125:1234–45.
Wang Q, Chen Z, Guo J, Peng X, Zheng Z, Chen H, Liu H, Ma Y, Zhu J. Atorvastatin-induced tolerogenic dendritic cells improve cardiac remodeling by suppressing TLR-4/NF-kappaB activation after myocardial infarction. Inflamm Res. 2023;72:13–25.
Chen S, Wang K, Fan Z, Zhou T, Li R, Zhang B, et al. Modulation of anti-cardiac fibrosis immune responses by changing M2 macrophages into M1 macrophages. Mol Med. 2024;30:88.
Ikeda M, Ide T, Matsushima S, Ikeda S, Okabe K, Ishikita A, et al. Immunomodulatory cell therapy using αGalCer-pulsed dendritic cells ameliorates heart failure in a murine dilated cardiomyopathy model. Circ Heart Fail. 2022;15: e009366.
Amrute JM, Luo X, Penna V, Yang S, Yamawaki T, Hayat S, et al. Targeting immune-fibroblast cell communication in heart failure. Nature. 2024;635:423–33.
Zhang Q, Dai J, Liu T, Rao W, Li D, Gu Z, Huang L, Wang J, Hou X. Targeting cardiac fibrosis with chimeric antigen receptor-engineered cells. Mol Cell Biochem. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11010-024-05134-6.
Maejima Y, Okada H, Haraguchi G, Onai Y, Kosuge H, Suzuki J, Isobe M. Telmisartan, a unique ARB, improves left ventricular remodeling of infarcted heart by activating PPAR gamma. Lab Invest. 2011;91:932–44.
Rurik JG, Fernández POM, Li L, Shewale SV. CAR T cells produced in vivo to treat cardiac injury. Science. 2022;375:91–6.
Gao Z, Yan L, Meng J, Lu Z, Ge K, Jiang Z, Feng T, Wang H, Liu C, Tang J, Zhang H. Targeting cardiac fibrosis with chimeric antigen receptor macrophages. Cell Discov. 2024;10:86.
Zhao X, Qin Y, Li B, Wang Y, Liu J, Wang B, et al. Genetically engineered biomimetic ATP-responsive nanozyme for the treatment of cardiac fibrosis. J Nanobiotechnol. 2025;23:10.
Zhang Y, Diao HT, Leng MY, Wu YZ, Huang BY, Li X, et al. YTHDF3-mediated FLCN/cPLA2 axis improves cardiac fibrosis via suppressing lysosomal function. Acta Pharmacol Sin. 2025. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41401-024-01425-2.
Zannad F, Gattis Stough W, Rossignol P, Bauersachs J, McMurray JJ, Swedberg K, et al. Mineralocorticoid receptor antagonists for heart failure with reduced ejection fraction: integrating evidence into clinical practice. Eur Heart J. 2012;33:2782–95.
Kamal NH, Heikal LA, Abdallah OY. The future of cardiac repair: a review on cell-free nanotherapies for regenerative myocardial infarction. Drug Deliv Transl Res. 2025. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13346-024-01763-y.
Jardin B, Epstein JA. Emerging mRNA therapies for cardiac fibrosis. Am J Physiol Cell Physiol. 2024;326:C107-c111.
Wang W, Li Y, Zhang C, Zhou H, Li C, Cheng R, Chen X, Pu Y, Chen Y. Small extracellular vesicles from young healthy human plasma inhibit cardiac fibrosis after myocardial infarction via miR-664a-3p targeting SMAD4. Int J Nanomed. 2025;20:557–79.
Schaefer AK, Kiss A, Oszwald A, Nagel F, Acar E, Aliabadi-Zuckermann A, et al. Single donor infusion of S-nitroso-human-serum-albumin attenuates cardiac isograft fibrosis and preserves myocardial micro-RNA-126-3p in a murine heterotopic heart transplant model. Transpl Int. 2022;35:10057.
Burrage MK, Cheshire C, Hey CY, Azam S, Watson WD, Bhagra S, et al. Comparing cardiac mechanics and myocardial fibrosis in DBD and DCD heart transplant recipients. J Card Fail. 2023;29:834–40.
Chi J, Wang L, Zhang X, Fu Y, Liu Y, Chen W, Liu W, Shi Z, Yin X. Cyclosporin A induces autophagy in cardiac fibroblasts through the NRP-2/WDFY-1 axis. Biochimie. 2018;148:55–62.
Abdullah CS, Li Z, Wang X, Jin ZQ. Depletion of T lymphocytes ameliorates cardiac fibrosis in streptozotocin-induced diabetic cardiomyopathy. Int Immunopharmacol. 2016;39:251–64.
Rurik JG, Aghajanian H, Epstein JA. Immune cells and immunotherapy for cardiac injury and repair. Circ Res. 2021;128:1766–79.
Masurkar N, Bouvet M, Logeart D, Jouve C, Drame F, Claude O, et al. Novel cardiokine GDF3 predicts adverse fibrotic remodeling after myocardial infarction. Circulation. 2023;147:498–511.
Acknowledgements
All figures were created on BioRender.com.
Funding
This work was supported by the National Natural Science Foundation of China (82172168, 82372202).
Author information
Authors and Affiliations
Contributions
XMH contributed to the conception of the review. DL performed the literature search. WTR drafted the main manuscript, and produced the figures. QHZ, TBL, ZYG, LH, JJD and JYW revised and corrected the manuscript. All authors reviewed the manuscript and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
All authors read and approved the final manuscript.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Rao, W., Li, D., Zhang, Q. et al. Complex regulation of cardiac fibrosis: insights from immune cells and signaling pathways. J Transl Med 23, 242 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06260-5
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06260-5